Background Cofilin is a member of the actin depolymerizing factor

Background Cofilin is a member of the actin depolymerizing factor Rabbit Polyclonal to MRPS24. (ADF)/cofilin family which regulates actin dynamics. of PP1 and PP2A. Only dephosphorylated cofilin binds to G-actin and translocates to mitochondria during AITC-mediated apoptosis. Mechanistic study revealed that interruption of ROCK1/PTEN/PI3K signaling pathway plays a critical role in AITC-mediated dephosphorylation and mitochondrial translocation of cofilin and apoptosis. Our study also showed that AITC-mediated inhibition of tumor growth of mouse leukemia xenograft model is usually in association with dephosphorylation of cofilin. Conclusions These findings support a model in which induction of apoptosis by AITC stems primarily from activation of ROCK1 and PTEN and inactivation of PI3K leading in turn to activation of PP1 and PP2A resulting in dephosphorylation of cofilin which binds to G-actin and translocates Fmoc-Lys(Me3)-OH chloride to mitochondria culminating in the dysfunction of mitochondria release of cytochrome c and apoptosis. and in several tumor xenograft models results indicate that dephosphorylation of cofilin may contribute to AITC-mediated inhibitory effects on tumor growth of U937 Fmoc-Lys(Me3)-OH chloride xenograft mouse model. These findings provide a novel mechanistic basis for AITC as a leukemia treatment strategy. Results AITC potently induces mitochondrial injury and apoptosis in transformed and primary human leukemia cells Flow cytometry analysis revealed that exposure of cells to 5 μM AITC for 24 h resulted in a moderate increase in mitochondrial injury (loss of △ψm) and apoptosis (Physique?1A). These events became apparent at 10 μM and very extensive at 20 μM concentrations. A time-course study of cells exposed to 20 μM AITC revealed a moderate increase in mitochondrial injury and apoptosis as early as 6 h after drug exposure. These events became apparent after 9 and 12 h of drug exposure and very extensive after 24 h of drug exposure (Physique?1A). Consistent with these findings the same AITC concentrations and exposure intervals caused cleavage/activation of caspase-9 and caspase-3 and degradation of PARP. These events were also accompanied by release of cytochrome c into the cytosolic fraction (Physique?1B). Physique 1 AITC selectively induces apoptosis and mitochondrial injury in transformed and primary human leukemia cells. U937 cells were treated without or with various concentrations of AITC for 24 h or treated with 20 μM AITC for different time intervals … To determine whether these events were restricted to myeloid leukemia cells parallel studies were performed in Jurkat and HL-60 leukemia cells. These cells exhibited apoptotic effects of AITC similar to those observed in U937 cells (Physique?1C). Also Jurkat and HL-60 cells exhibited comparable degrees of caspase-9 and -3 activation and PARP degradation and cytochrome c release (Physique?1D). To determine whether AITC could also trigger apoptosis in primary human leukemia cells primary leukemia cells isolated from 17 AML patients were treated without or with 20 μM AITC for 24 h after which apoptosis was determined by Annexin V/PI Fmoc-Lys(Me3)-OH chloride analysis. Exposure of these AML blasts to AITC resulted in marked increase in apoptosis (Physique?1E). Consistent with these findings treatment of leukemia blasts from 2 AML patients with AITC also resulted in cleavage/activation of caspase-9 and -3 degradation of PARP and release of cytochrome c (Physique?1F). In contrast AITC exerted little toxicity toward normal CD34+ bone marrow cells (Physique?1G). Taken together these findings suggest that AITC selectively induces mitochondrial injury and apoptosis in transformed and primary human leukemia cells but not in normal hematopoietic cells. Alteration of G/F-actin ratio and actin dynamics in response to AITC G/F-actin ratio is an indicator of the extent of actin dynamics and might be responsible for regulating apoptosis [5]. Fmoc-Lys(Me3)-OH chloride To understand the mechanism of AITC-mediated apoptosis through affecting actin dynamics we separated actin into G and F fractions and evaluated their relative content. Exposure of cells to AITC resulted in decrease in the polymerized F-actin and increase in the unpolymerized G-actin (Physique?2A). Consistent with these findings confocal microscopy showed that treating with AITC for different time intervals resulted in decrease in levels of F-actin and increase in levels of G-actin (Physique?2B). Such.